34 results on '"von Metzler I"'
Search Results
2. Incorporation of the bone marker carboxy-terminal telopeptide of type-1 collagen improves prognostic information of the International Staging System in newly diagnosed symptomatic multiple myeloma
- Author
-
Jakob, C, Sterz, J, Liebisch, P, Mieth, M, Rademacher, J, Goerke, A, Heider, U, Fleissner, C, Kaiser, M, von Metzler, I, Müller, C, and Sezer, O
- Published
- 2008
- Full Text
- View/download PDF
3. Bortezomib inhibits human osteoclastogenesis
- Author
-
von Metzler, I, Krebbel, H, Hecht, M, Manz, R A, Fleissner, C, Mieth, M, Kaiser, M, Jakob, C, Sterz, J, Kleeberg, L, Heider, U, and Sezer, O
- Published
- 2007
- Full Text
- View/download PDF
4. The proteasome inhibitor bortezomib stimulates osteoblastic differentiation of mesenchymal stem cells via upregulation of vitamin D receptor signaling: Implications for multiple myeloma: V619
- Author
-
Heider, U., Kaiser, M., Mieth, M., von Metzler, I., and Sezer, O.
- Published
- 2010
5. Erratum: Incorporation of the bone marker carboxy-terminal telopeptide of type-1 collagen improves prognostic information of the International Staging System in newly diagnosed symptomatic multiple myeloma
- Author
-
Jakob, C, Sterz, J, Liebisch, P, Mieth, M, Rademacher, J, Goerke, A, Heider, U, Fleissner, C, Kaiser, M, von Metzler, I, Müller, C, and Sezer, O
- Published
- 2008
- Full Text
- View/download PDF
6. NK Cell Anti-Tumor Efficacy in Multiple Myeloma Patients Before and After Autologous Stem Cell Transplantation.
- Author
-
Tognarelli, S., von Metzler, I., Rais, B., Jacobs, B., Bader, P., Serve, H., Mackensen, A., and Ullrich, E.
- Published
- 2017
- Full Text
- View/download PDF
7. CD8 + CD28 - regulatory T cells after induction therapy predict progression-free survival in myeloma patients: results from the GMMG-HD6 multicenter phase III study.
- Author
-
Kriegsmann K, Ton GNHQ, Awwad MHS, Benner A, Bertsch U, Besemer B, Hänel M, Fenk R, Munder M, Dürig J, Blau IW, Huhn S, Hose D, Jauch A, Mann C, Weinhold N, Scheid C, Schroers R, von Metzler I, Schieferdecker A, Thomalla J, Reimer P, Mahlberg R, Graeven U, Kremers S, Martens UM, Kunz C, Hensel M, Seidel-Glätzer A, Weisel KC, Salwender HJ, Müller-Tidow C, Raab MS, Goldschmidt H, Mai EK, and Hundemer M
- Subjects
- Humans, Male, Antineoplastic Combined Chemotherapy Protocols therapeutic use, CD8-Positive T-Lymphocytes immunology, Induction Chemotherapy, Prognosis, Progression-Free Survival, Survival Rate, CD28 Antigens, Multiple Myeloma mortality, Multiple Myeloma immunology, Multiple Myeloma drug therapy, Multiple Myeloma pathology, T-Lymphocytes, Regulatory immunology
- Published
- 2024
- Full Text
- View/download PDF
8. Predictors of early morbidity and mortality in newly diagnosed multiple myeloma: data from five randomized, controlled, phase III trials in 3700 patients.
- Author
-
Mai EK, Hielscher T, Bertsch U, Salwender HJ, Zweegman S, Raab MS, Munder M, Pantani L, Mancuso K, Brossart P, Beksac M, Blau IW, Dürig J, Besemer B, Fenk R, Reimer P, van der Holt B, Hänel M, von Metzler I, Graeven U, Müller-Tidow C, Boccadoro M, Scheid C, Dimopoulos MA, Hillengass J, Weisel KC, Cavo M, Sonneveld P, and Goldschmidt H
- Subjects
- Humans, Middle Aged, Morbidity, Risk Factors, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy
- Abstract
Early morbidity and mortality affect patient outcomes in multiple myeloma. Thus, we dissected the incidence and causes of morbidity/mortality during induction therapy (IT) for newly diagnosed multiple myeloma (NDMM), and developed/validated a predictive risk score. We evaluated 3700 transplant-eligible NDMM patients treated in 2005-2020 with novel agent-based triplet/quadruplet IT. Primary endpoints were severe infections, death, or a combination of both. Patients were divided in a training (n = 1333) and three validation cohorts (n = 2367). During IT, 11.8%, 1.8%, and 12.5% of patients in the training cohort experienced severe infections, death, or both, respectively. Four major, baseline risk factors for severe infection/death were identified: low platelet count (<150/nL), ISS III, higher WHO performance status (>1), and age (>60 years). A risk score (1 risk factor=1 point) stratified patients in low (39.5%; 0 points), intermediate (41.9%; 1 point), and high (18.6%; ≥2 points) risk. The risk for severe infection/death increased from 7.7% vs. 11.5% vs. 23.3% in the low- vs. intermediate- vs. high-risk groups (p < 0.001). The risk score was independently validated in three trials incorporating quadruplet IT with an anti-CD38 antibody. Our analyses established a robust and easy-to-use score to identify NDMM patients at risk of severe infection/death, covering the latest quadruplet induction therapies. Trial registrations: HOVON-65/GMMG-HD4: EudraCT No. 2004-000944-26. GMMG-MM5: EudraCT No. 2010-019173-16. GMMG-HD6: NCT02495922. EMN02/HOVON-95: NCT01208766. GMMG-HD7: NCT03617731., (© 2023. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. Elotuzumab, lenalidomide, bortezomib, dexamethasone, and autologous haematopoietic stem-cell transplantation for newly diagnosed multiple myeloma (GMMG-HD6): results from a randomised, phase 3 trial.
- Author
-
Mai EK, Goldschmid H, Miah K, Bertsch U, Besemer B, Hänel M, Krzykalla J, Fenk R, Schlenzka J, Munder M, Dürig J, Blau IW, Huhn S, Hose D, Jauch A, Kunz C, Mann C, Weinhold N, Scheid C, Schroers R, von Metzler I, Schieferdecker A, Thomalla J, Reimer P, Mahlberg R, Graeven U, Kremers S, Martens UM, Kunz C, Hensel M, Benner A, Seidel-Glätzer A, Weisel KC, Raab MS, and Salwender HJ
- Subjects
- Adult, Male, Female, Humans, Lenalidomide adverse effects, Bortezomib adverse effects, Dexamethasone adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Transplantation, Autologous, Multiple Myeloma drug therapy, Multiple Myeloma diagnosis, Hematopoietic Stem Cell Transplantation adverse effects, Pneumonia etiology, Sepsis chemically induced, Sepsis drug therapy, Antibodies, Monoclonal, Humanized
- Abstract
Background: The aim of this trial was to investigate the addition of the anti-SLAMF7 monoclonal antibody elotuzumab to lenalidomide, bortezomib, and dexamethasone (RVd) in induction and consolidation therapy as well as to lenalidomide maintenance treatment in transplant-eligible patients with newly diagnosed multiple myeloma., Methods: GMMG-HD6 was a phase 3, randomised trial conducted at 43 main trial sites and 26 associated trial sites throughout Germany. Adult patients (aged 18-70 years) with previously untreated, symptomatic multiple myeloma, and a WHO performance status of 0-3, with 3 being allowed only if caused by myeloma disease and not by comorbid conditions, were randomly assigned 1:1:1:1 to four treatment groups. Induction therapy consisted of four 21-day cycles of RVd (lenalidomide 25 mg orally on days 1-14; bortezomib 1·3 mg/m
2 subcutaneously on days 1, 4, 8, and 11]; and dexamethasone 20 mg orally on days 1, 2, 4, 5, 8, 9, 11, 12, and 15 for cycles 1-2) or, RVd induction plus elotuzumab (10 mg/kg intravenously on days 1, 8, and 15 for cycles 1-2, and on days 1 and 11 for cycles 3-4; E-RVd). Autologous haematopoietic stem-cell transplantation was followed by two 21-day cycles of either RVd consolidation (lenalidomide 25 mg orally on days 1-14; bortezomib 1·3 mg/m2 subcutaneously on days 1, 8, and 15; and dexamethasone 20 mg orally on days 1, 2, 8, 9, 15, and 16) or elotuzumab plus RVd consolidation (with elotuzumab 10 mg/kg intravenously on days 1, 8, and 15) followed by maintenance with either lenalidomide (10 mg orally on days 1-28 for cycles 1-3; thereafter, up to 15 mg orally on days 1-28; RVd/R or E-RVd/R group) or lenalidomide plus elotuzumab (10 mg/kg intravenously on days 1 and 15 for cycles 1-6, and on day 1 for cycles 7-26; RVd/E-R or E-RVd/E-R group) for 2 years. The primary endpoint was progression-free survival analysed in a modified intention-to-treat (ITT) population. Safety was analysed in all patients who received at least one dose of trial medication. This trial is registered with ClinicalTrials.gov, NCT02495922, and is completed., Findings: Between June 29, 2015, and on Sept 11, 2017, 564 patients were included in the trial. The modified ITT population comprised 559 (243 [43%] females and 316 [57%] males) patients and the safety population 555 patients. After a median follow-up of 49·8 months (IQR 43·7-55·5), there was no difference in progression-free survival between the four treatment groups (adjusted log-rank p value, p=0·86), and 3-year progression-free survival rates were 69% (95% CI 61-77), 69% (61-76), 66% (58-74), and 67% (59-75) for patients treated with RVd/R, RVd/E-R, E-RVd/R, and E-RVd/E-R, respectively. Infections (grade 3 or worse) were the most frequently observed adverse event in all treatment groups (28 [20%] of 137 for RVd/R; 32 [23%] of 138 for RVd/E-R; 35 [25%] of 138 for E-RVd/R; and 48 [34%] of 142 for E-RVd/E-R). Serious adverse events (grade 3 or worse) were observed in 68 (48%) of 142 participants in the E-RVd/E-R group, 53 (39%) of 137 in the RVd/R, 53 (38%) of 138 in the RVd/E-R, and 50 (36%) of 138 in the E-RVd/R (36%) group. There were nine treatment-related deaths during the study. Two deaths (one sepsis and one toxic colitis) in the RVd/R group were considered lenalidomide-related. One death in the RVd/E-R group due to meningoencephalitis was considered lenalidomide and elotuzumab-related. Four deaths (one pulmonary embolism, one septic shock, one atypical pneumonia, and one cardiovascular failure) in the E-RVd/R group and two deaths (one sepsis and one pneumonia and pulmonary fibrosis) in the E-RVd/E-R group were considered related to lenalidomide or elotuzumab, or both., Interpretation: Addition of elotuzumab to RVd induction or consolidation and lenalidomide maintenance in patients with transplant-eligible newly diagnosed multiple myeloma did not provide clinical benefit. Elotuzumab-containing therapies might be reserved for patients with relapsed or refractory multiple myeloma., Funding: Bristol Myers Squibb/Celgene and Chugai., Competing Interests: Declaration of interests EKM reports consulting or advisory role with Bristol Myers Squibb (BMS)/Celgene, GlaxoSmithKline, Janssen-Cilag, Sanofi Aventis, Stemline, and Takeda; honoraria from BMS/Celgene, GlaxoSmithKline, Janssen-Cilag, Sanofi Aventis, Stemline, and Takeda; research funding from Sanofi Aventis; and travel accommodation and expenses from BMS/Celgene, GlaxoSmithKline, Janssen-Cilag, Sanofi Aventis, Stemline, and Takeda. HG reports support for the present manuscript from BMS/Celgene, Chugai, and HD6 funding; consulting or advisory role with Amgen, BMS, Janssen, Sanofi, and Adaptive Biotechnology; honoraria from Amgen, BMS, Chugai, GlaxoSmithKline, Janssen, Novartis, Sanofi, and Pfizer; research funding from Amgen, BMS, Celgene, GlycoMimetics, GlaxoSmithKline, Heidelberg Pharma, Hoffmann-La Roche, Karyopharm, Janssen, Incyte Corporation, Millenium Pharmaceuticals, Molecular Partners, Merck Sharp and Dohme, MorphoSys, Pfizer, Sanofi, Takeda, and Novartis; travel accommodations and expenses from Amgen, BMS, GlaxoSmithKline, Janssen, Novartis, Sanofi, and Pfizer; and grants or provision of Investigational Medicinal Products from Amgen, Array Biopharma/Pfizer, BMS/Celgene, Chugai, Dietmar-Hopp-Foundation, Janssen, Johns Hopkins University, Mundipharma, and Sanofi. MHä reports consulting or advisory role with Novartis, BMS/Celgene, Gilead Sciences, Sanofi/Aventis, Roche, Amgen, SOBI, Janssen, Takeda, GlaxoSmithKline, Jazz Pharmaceuticals, Bayer Vital, and BMS. RF reports Honoraria from Amgen, BMS/Celgene, Janssen, Sanofi, and Takeda; and travel accommodations and expenses from BMS/Celgene, Janssen, and GSK. MM reports consulting or Advisory Role with Janssen, BMS, Abbvie, Sanofi, GlaxoSmithKline, Oncopeptides, Takeda, and Stemline; honoraria from Amgen, BMS, GlaxoSmithKline, Janssen, and Takeda; and travel accommodations and expenses from Amgen. JD reports honoraria from Sanofi, BMS, Janssen, AstraZeneca, Beigene; and travel accommodation and expenses from Amgen, BMS, Beigene, and Amgen. CM reports Consulting or Advisory Role from Celgene, BMS, and Janssen. CS reports consulting or advisory role from BMS, GlaxoSmithKline, Janssen, Pfizer, Roche, and Takeda; honoraria from Amgen, BMS/GlaxoSmithKline, Janssen, MSD, Novartis, Roche, Sanofi, and Takeda; research funding from Janssen, and Takeda; and travel accommodation and expenses from BMS, Janssen, Sanofi Aventis, and Takeda. RS reports consulting or advisory role with BMS/Celgene, GlaxoSmithKline, Janssen, Kite/Gilead, and Sanofi; and honoraria from Amgen, BMS/Celgene, GlaxoSmithKline, Janssen, Kite/Gilead, Sanofi, and Takeda. IvM reports consulting or advisory role with Sanofi, Amgen, Janssen, Takeda, Stemline, GSK, BMS, Oncopeptides, Pfizer, and AstraZeneca. PR reports honoraria from BMS and travel accommodation and expenses from BMS. UG reports consulting or advisory role with Amgen, Boehringer Ingelheim, BMS, Celtrion, Ipsen, Sanofi, and MSD; and honoraria from Amgen, AstraZeneca, and Novartis. SK reports travel accommodation and expenses from AbbVie. UMM reports consulting or advisory role with Sanofi-Aventis, BMS, Roche, GSK, Novartis, Pierre Fabre, MSD, and Guardant Health; and travel accommodation and expenses from Pierre Fabre, Ipsen, and Janssen. CK reports travel accommodation and expenses from European Hematology Association. KCW reports consulting or advisory role with Abbvie, Amgen, Adaptive Biotech, BMS/Celgene, BeiGene, Janssen, GlaxoSmithKline, Karyopharm, Oncopeptides, Pfizer, Roche Pharma, Sanofi, Takeda, and Menarini; honoraria from Abbvie, Amgen, Adaptive Biotech, Astra Zeneca, BMS/Celgene, BeiGene, Janssen, GlaxoSmithKline, Karyopharm, Novartis, Oncopeptides, Pfizer, Roche Pharma, Sanofi, Stemline, Takeda, and Menarini; and research funding from Abbvie, Amgen, BMS/Celgene, Janssen, GlaxoSmithKline, Sanofi, and Takeda. MSR reports consulting or advisory role with BMS, Amgen, GSK, Janssen, Sanofi, Pfizer, AbbVie, Novartis, and Roche; research funding from Sanofi; travel accommodation and expenses from BMS, AbbVie, Janssen, Sanofi, GSK; Honoraria from BMS, Janssen, GSK, AbbVie, and Sanofi; and receipt of equipment, materials, drugs, medical writing, gifts or other services from Novartis, Sanofi. HJS reports consulting or advisory role with Amgen, AstraZeneca, BMS/Celgene, Genzyme, GSK, Janssen Cilag, Oncopeptides, Pfizer, Sanofi, and Stemline; honoraria from Abbvie, Amgen, AstraZeneca, BMS/Celgene, Genzyme, GSK, Janssen Cilag, Oncopeptides, Pfizer, Roche, Sanofi, Stemline, and Takeda; and travel accommodation and expenses from Amgen, BMS/Celgene, Janssen Cilag, and Sanofi. All other authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
10. Cytokine-responsive T- and NK-cells portray SARS-CoV-2 vaccine-responders and infection in multiple myeloma patients.
- Author
-
Enssle JC, Campe J, Moter A, Voit I, Gessner A, Yu W, Wolf S, Steffen B, Serve H, Bremm M, Huenecke S, Lohoff M, Vehreschild M, Rabenau HF, Widera M, Ciesek S, Oellerich T, Imkeller K, Rieger MA, von Metzler I, and Ullrich E
- Subjects
- Humans, COVID-19 Vaccines, Cytokines, Leukocytes, Mononuclear, SARS-CoV-2, Vaccination, Multiple Myeloma therapy, COVID-19
- Abstract
Patients with multiple myeloma (MM) routinely receive mRNA-based vaccines to reduce COVID-19-related mortality. However, whether disease- and therapy-related alterations in immune cells and cytokine-responsiveness contribute to the observed heterogeneous vaccination responses is unclear. Thus, we analyzed peripheral blood mononuclear cells from patients with MM during and after SARS-CoV-2 vaccination and breakthrough infection (BTI) using combined whole-transcriptome and surface proteome single-cell profiling with functional serological and T-cell validation in 58 MM patients. Our results demonstrate that vaccine-responders showed a significant overrepresentation of cytotoxic CD4
+ T- and mature CD38+ NK-cells expressing FAS+ /TIM3+ with a robust cytokine-responsiveness, such as type-I-interferon-, IL-12- and TNF-α-mediated signaling. Patients with MM experiencing BTI developed strong serological and cellular responses and exhibited similar cytokine-responsive immune cell patterns as vaccine-responders. This study can expand our understanding of molecular and cellular patterns associated with immunization responses and may benefit the design of improved vaccination strategies in immunocompromised patients., (© 2023. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
11. The MYC-Regulated RNA-Binding Proteins hnRNPC and LARP1 Are Drivers of Multiple Myeloma Cell Growth and Disease Progression and Negatively Predict Patient Survival.
- Author
-
Seibert M, Koschade SE, Stolp V, Häupl B, Wempe F, Serve H, Kurrle N, Schnütgen F, and von Metzler I
- Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder in which the MYC oncogene is frequently dysregulated. Due to its central role, MYC has been proposed as a drug target; however, the development of a clinically applicable molecule modulating MYC activity remains an unmet challenge. Consequently, an alternative is the development of therapeutic options targeting proteins located downstream of MYC. Therefore, we aimed to identify undescribed MYC-target proteins in MM cells using Stable Isotope Labeling with Amino Acids in Cell Culture (SILAC) and mass spectrometry. We revealed a cluster of proteins associated with the regulation of translation initiation. Herein, the RNA-binding proteins Heterogeneous Nuclear Ribonucleoprotein C (hnRNPC) and La Ribonucleoprotein 1 (LARP1) were predominantly downregulated upon MYC depletion. CRISPR-mediated knockout of either hnRNPC or LARP1 in conjunction with redundant LARP family proteins resulted in a proliferative disadvantage for MM cells. Moreover, high expression levels of these proteins correlate with high MYC expression and with poor survival and disease progression in MM patients. In conclusion, our study provides valuable insights into MYC's role in translation initiation by identifying hnRNPC and LARP1 as proliferation drivers of MM cells and as both predictive factors for survival and disease progression in MM patients., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
12. Comparison of bone marrow and peripheral blood aberrant plasma cell assessment by NGF in patients with MM.
- Author
-
Kriegsmann K, Manta C, Schwab R, Mai EK, Raab MS, Salwender HJ, Fenk R, Besemer B, Dürig J, Schroers R, von Metzler I, Hänel M, Mann C, Asemissen AM, Heilmeier B, Bertsch U, Huhn S, Müller-Tidow C, Goldschmidt H, and Hundemer M
- Subjects
- Humans, Bone Marrow Cells, Bone Marrow, Plasma Cells
- Published
- 2023
- Full Text
- View/download PDF
13. Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone as induction therapy for newly diagnosed, transplantation-eligible patients with multiple myeloma (GMMG-HD7): part 1 of an open-label, multicentre, randomised, active-controlled, phase 3 trial.
- Author
-
Goldschmidt H, Mai EK, Bertsch U, Fenk R, Nievergall E, Tichy D, Besemer B, Dürig J, Schroers R, von Metzler I, Hänel M, Mann C, Asemissen AM, Heilmeier B, Weinhold N, Huhn S, Kriegsmann K, Luntz SP, Holderried TAW, Trautmann-Grill K, Gezer D, Klaiber-Hakimi M, Müller M, Khandanpour C, Knauf W, Scheid C, Munder M, Geer T, Riesenberg H, Thomalla J, Hoffmann M, Raab MS, Salwender HJ, and Weisel KC
- Subjects
- Male, Humans, Female, Middle Aged, Lenalidomide therapeutic use, Bortezomib adverse effects, Induction Chemotherapy, Dexamethasone, Antineoplastic Combined Chemotherapy Protocols adverse effects, Multiple Myeloma therapy
- Abstract
Background: Anti-CD38 monoclonal antibodies have consistently shown increased efficacy when added to standard of care for patients with multiple myeloma. We aimed to assess the efficacy of isatuximab in addition to lenalidomide, bortezomib, and dexamethasone in patients with newly diagnosed transplantation-eligible multiple myeloma., Methods: This open-label, multicentre, randomised, active-controlled, phase 3 trial was done at 67 academic and oncology practice centres in Germany. This study is ongoing and divided into two parts; herein, we report results from part 1. Eligible patients were aged 18-70 years; had a confirmed diagnosis of untreated multiple myeloma requiring systemic treatment and a WHO performance status of 0-2; and were eligible for induction therapy, high-dose melphalan and autologous haematopoietic stem-cell transplantation, and maintenance treatment. Patients were randomly assigned (1:1) to receive three 42-day cycles of induction therapy either with isatuximab plus lenalidomide, bortezomib, and dexamethasone (isatuximab group) or lenalidomide, bortezomib, and dexamethasone alone (control group) using a web-based system and permuted blocks. Patients in both groups received lenalidomide (25 mg orally on days 1-14 and 22-35), bortezomib (1·3 mg/m
2 subcutaneously on days 1, 4, 8, 11, 22, 25, 29, and 32), and dexamethasone (20 mg orally on days 1-2, 4-5, 8-9, 11-12, 15, 22-23, 25-26, 29-30, and 32-33). Isatuximab was given as 10 mg/kg intravenously on days 1, 8, 15, 22, and 29 of cycle 1 and on days 1, 15, and 29 of cycles 2 and 3. The primary endpoint was minimal residual disease (MRD) negativity assessed by flow cytometry, in the intention-to-treat (ITT) population. This study is registered with ClinicalTrials.gov, NCT03617731., Findings: Between Oct 23, 2018, and Sep 22, 2020, 660 patients were included in the ITT analysis (331 in the isatuximab group and 329 in the control group). 654 (99%) patients were White, two were African, one was Arabic, and three were Asian. 250 (38%) were women and 410 (62%) were men. The median age was 59 years (IQR 54-64). MRD negativity after induction therapy was reached in 166 (50%) patients in the isatuximab group versus 117 (36%) in the control group (OR 1·82 [95% CI 1·33-2·48]; p=0·00017). Median follow-up time from start to end of induction therapy was 125 days (IQR 125-131) versus 125 days (125-132). At least one grade 3 or 4 adverse event occurred in 208 (63%) of 330 patients versus 199 (61%) of 328 patients. Neutropenia of grade 3 or 4 occurred in 77 (23%) versus 23 (7%) patients and infections of grade 3 or 4 occurred in 40 (12%) versus 32 (10%) patients. Among 12 deaths during induction therapy, one death due to septic shock in the isatuximab group and four deaths (one cardiac decompensation, one hepatic and renal failure, one cardiac arrest, and one drug-induced enteritis) in the control group were considered treatment-related., Interpretation: Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone for induction therapy improved rates of MRD negativity with no new safety signals in patients with newly diagnosed transplantation-eligible multiple myeloma., Funding: Sanofi and Bristol Myers Squibb (Celgene)., Competing Interests: Declaration of interests HG reports financial and trial medication support for this GMMG-HD7 trial from Bristol Myers Squibb and Sanofi; consulting from Adaptive Biotechnology, Amgen, Bristol Myers Squibb, and Janssen; honoraria from Amgen, Bristol Myers Squibb, Celgene, Chugai, GlaxoSmithKline, Janssen, and Novartis; research funding from Amgen, Bristol Myers Squibb, Celgene, Chugai, Incyte, Janssen, Molecular Partner, MSD, Mundipharma, and Novartis; grants from Amgen, Celgene, Chugai, Janssen, and Johns Hopkins University. EKM reports consulting or advisory role, honoraria, research funding, travel accommodation, and expenses from Bristol Myers Squibb (Celgene), GlaxoSmithKline, Janssen-Cilag, Sanofi, Stemline, and Takeda. RF reports consulting or advisory role, honoraria, travel accommodation, and expenses from Amgen, Bristol Myers Squibb (Celgene), GlaxoSmithKline, Janssen, and Takeda. BB reports honoraria from Amgen, GlaxoSmithKline, Janssen, Sanofi, and Takeda; and travel accommodation, and expanses from Janssen. JD reports honoraria from Celgene, Janssen, and Roche; advisory role, travel accommodation, and expenses from Amgen, AstraZeneca, Abbvie, Bristol Myers Squibb, Celgene, Janssen, and Takeda; and speakers bureau from Celgene and Janssen. RS reports consulting for Bristol Myers Squibb, Gilead (Kite), Janssen, and Novartis; and honoraria from Bristol Myers Squibb, Gilead (Kite), Janssen, Roche, and Sanofi. IvM reports consulting for Amgen, AstraZeneca, Bristol Myers Squibb, GlaxoSmithKline, Janssen, Pfizer, Sanofi, and Takeda; honoraria from Sanofi; and travel accommodation and expenses from Takeda. MHa reports consulting for Amgen, Bayer Vital, Celgene, Gilead, GlaxoSmithKline, Jazz Pharmaceuticals, Novartis, Pfizer, Roche, and Takeda; and honoraria from Novartis. CM reports consulting for Celgene. AMA reports honoraria from Bristol Myers Squibb (Celgene), GlaxoSmithKline, and Pfizer. BH reports consulting for Sanofi-Aventis. NW reports an advisory role for Glaxo Smith Kline; and research Funding from Bristol Myers Squibb. KK reports honoraria from Sanofi. TAWH reports a consulting or advisory role for Celgene, Gilead Sciences, GlaxoSmithKline, Jazz Pharmaceuticals, Novartis, and Sanofi; speakers bureau for Amgen and MSD; and travel accommodation and expenses from AbbVie, Daiichi Sankyo, Eurocept Pharmaceuticals, Janssen, Medac, and Therakos. KT-G reports consulting, an advisory role, honoraria, travel accommodation, and expenses from GlaxoSmithKline, Novartis, and Takeda. DG reports consulting and honoraria from Amgen, Bristol Myers Squibb, Celgene, and Takeda. MK-H reports honoraria from Amgen, Bristol Myers Squibb, Janssen-Cilag, Roche, and Takeda; and an advisory role for Amgen, Bristol Myers Squibb, Janssen, and Sanofi. CK reports honoraria from AstraZeneca, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Pfizer, Sanofi, and Takeda; and research funding from AstraZeneca and Sanofi. WK reports consulting for AstraZeneca, BeiGene, and Janssen; honoraria from AbbVie, Amgen, AstraZeneca, BeiGene, Bristol Myers Squibb, Celgene, Janssen, and Sanofi. CS reports honoraria from AbbVie, Amgen, Bristol Myers Squibb, Glaxo Smith Kline, Janssen, Pfizer, Oncopeptides, Sanofi, and Takeda; travel accommodation and expanses from Bristol Myers Squibb, Janssen, Novartis, and Takeda; and research funding from Janssen, Novartis, and Takeda. MMun reports consulting for AbbVie, Bristol Myers Squibb, Glaxo Smith Kline, Janssen, Sanofi, and Takeda; honoraria from Amgen, Bristol Myers Squibb, Janssen, and Takeda; research funding from Incyte; and travel accommodation and expenses from Amgen. MHo reports consulting for Sanofi-Aventis. MSR reports consulting or an advisory role for AbbVie, Amgen, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Novartis, Roche, and Sanofi; honoraria from AbbVie and GlaxoSmithKline; and research funding from Novartis and Sanofi. HJS reports honoraria and an advisory role from AbbVie, Amgen, Bristol Myers Squibb (Celgene), Chugai, GlaxoSmithKline, Janssen-Cilag, Oncopeptides, Pfizer, Roche, Sanofi, Sebia, TAD Pharma, and Takeda; travel accommodation and expenses from Amgen, Bristol Myers Squibb (Celgene), GlaxoSmithKline, Janssen-Cilag, and Sanofi. KCW reports consulting for AbbVie, Adaptive Biotech, Amgen, AstraZeneca, BeiGene, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Karyopharm, Novartis, Oncopeptides, Pfizer, Roche, Sanofi, Stemline, and Takeda; honoraria from AbbVie, Adaptive Biotech, Amgen, AstraZeneca, BeiGene, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Karyopharm, Novartis, Oncopeptides, Pfizer, Roche, Sanofi, Stemline, and Takeda; research funding from Amgen, Bristol Myers Squibb, Celgene, Glaxo Smith Kline, Janssen, and Sanofi; travel accommodation and expenses from AbbVie, Adaptive Biotech, Amgen, Bristol Myers Squibb, Celgene, Janssen, Glaxo Smith Kline, Karyopharm, Oncopeptides, Roche, Sanofi, Stemline, and Takeda. All other authors declare no competing interests., (Copyright © 2022 Elsevier Ltd. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
14. Mitochondrial apoptosis: facilitator of NK cell-mediated immunotherapy.
- Author
-
Ullrich E, Vogler M, and von Metzler I
- Subjects
- Apoptosis genetics, Immunotherapy, Killer Cells, Natural
- Published
- 2022
- Full Text
- View/download PDF
15. Enhanced but variant-dependent serological and cellular immune responses to third-dose BNT162b2 vaccination in patients with multiple myeloma.
- Author
-
Enssle JC, Campe J, Büchel S, Moter A, See F, Grießbaum K, Rieger MA, Wolf S, Ballo O, Steffen B, Serve H, Rabenau HF, Widera M, Bremm M, Huenecke S, Ciesek S, von Metzler I, and Ullrich E
- Subjects
- BNT162 Vaccine, Humans, Immunity, Cellular, Vaccination, Multiple Myeloma
- Abstract
Competing Interests: Declaration of interests S.C. received honoraria for advising Pfizer/BioNTech. I.v.M. received honoraria for advising Pfizer, Sanofi, BMS, GSK, Amgen, Janssen, Takeda, and AstraZeneca. E.U. received honoraria for advising Phialogics and BMS. Other authors declare no conflict of interest to the present study.
- Published
- 2022
- Full Text
- View/download PDF
16. CRISPR-Cas9 based gene editing of the immune checkpoint NKG2A enhances NK cell mediated cytotoxicity against multiple myeloma.
- Author
-
Bexte T, Alzubi J, Reindl LM, Wendel P, Schubert R, Salzmann-Manrique E, von Metzler I, Cathomen T, and Ullrich E
- Subjects
- CRISPR-Cas Systems genetics, Gene Editing, Humans, Killer Cells, Natural metabolism, Multiple Myeloma genetics, Multiple Myeloma therapy, NK Cell Lectin-Like Receptor Subfamily C genetics, NK Cell Lectin-Like Receptor Subfamily C metabolism
- Abstract
Natural Killer (NK) cells are known for their high intrinsic cytotoxic capacity, and the possibility to be applied as 'off-the-shelf' product makes them highly attractive for cell-based immunotherapies. In patients with multiple myeloma (MM), an elevated number of NK cells has been correlated with higher overall-survival rate. However, NK cell function can be impaired by upregulation of inhibitory receptors, such as the immune checkpoint NKG2A. Here, we developed a CRISPR-Cas9-based gene editing protocol that allowed us to knockout about 80% of the NKG2A-encoding killer cell lectin like receptor C1 ( KLRC1 ) locus in primary NK cells. In-depth phenotypic analysis confirmed significant reduction in NKG2A protein expression. Importantly, the KLRC1- edited NK cells showed significantly increased cytotoxicity against primary MM cells isolated from a small cohort of patients, and maintained the NK cell-specific cytokine production. In conclusion, KLRC1 -editing in primary NK cells has the prospect of overcoming immune checkpoint inhibition in clinical applications., Competing Interests: No potential conflict of interest was reported by the author(s)., (© 2022 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2022
- Full Text
- View/download PDF
17. COVID-19 in multiple-myeloma patients: cellular and humoral immunity against SARS-CoV-2 in a short- and long-term view.
- Author
-
von Metzler I, Campe J, Huenecke S, Raab MS, Goldschmidt H, Schubert R, Rabenau HF, Ciesek S, Serve H, and Ullrich E
- Subjects
- Antibodies, Neutralizing immunology, Humans, Immunoglobulin G immunology, Male, Middle Aged, COVID-19 immunology, Immunity, Cellular immunology, Immunity, Humoral immunology, Multiple Myeloma immunology, Multiple Myeloma virology, SARS-CoV-2 immunology
- Abstract
Multiple myeloma patients are often treated with immunomodulatory drugs, proteasome inhibitors, or monoclonal antibodies until disease progression. Continuous therapy in combination with the underlying disease frequently results in severe humoral and cellular immunodeficiency, which often manifests in recurrent infections. Here, we report on the clinical management and immunological data of three multiple-myeloma patients diagnosed with COVID-19. Despite severe hypogammaglobulinemia, deteriorated T cell counts, and neutropenia, the patients were able to combat COVID-19 by balanced response of innate immunity, strong CD8+ and CD4+ T cell activation and differentiation, development of specific T-cell memory subsets, and development of anti-SARS-CoV-2 type IgM and IgG antibodies with virus-neutralizing capacities. Even 12 months after re-introduction of lenalidomide maintenance therapy, antibody levels and virus-neutralizing antibody titers remained detectable, indicating persisting immunity against SARS-CoV-2. We conclude that in MM patients who tested positive for SARS-CoV-2 and were receiving active MM treatment, immune response assessment could be a useful tool to help guide decision-making regarding the continuation of anti-tumor therapy and supportive therapy. KEY MESSAGES: Immunosuppression due to multiple myeloma might not be the crucial factor that is affecting the course of COVID-19. In this case, despite pre-existing severe deficits in CD4+ T-cell counts and IgA und IgM deficiency, we noticed a robust humoral and cellular immune response against SARS-CoV-2. Evaluation of immune response and antibody titers in MM patients that were tested positive for SARS-CoV-2 and are on active MM treatment should be performed on a larger scale; the findings might affect further treatment recommendations for COVID-19, MM treatment re-introduction, and isolation measures., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. Identification of the Cysteine Protease Legumain as a Potential Chronic Hypoxia-Specific Multiple Myeloma Target Gene.
- Author
-
Clees AS, Stolp V, Häupl B, Fuhrmann DC, Wempe F, Seibert M, Weber S, Banning A, Tikkanen R, Williams R, Brüne B, Serve H, Schnütgen F, von Metzler I, and Kurrle N
- Subjects
- Basic Helix-Loop-Helix Transcription Factors metabolism, CRISPR-Cas Systems genetics, Cell Line, Tumor, Cell Proliferation genetics, Gene Expression Regulation, Neoplastic, Hexokinase metabolism, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Lactate Dehydrogenase 5 metabolism, Proteome metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction genetics, Up-Regulation genetics, Cysteine Endopeptidases genetics, Molecular Targeted Therapy, Multiple Myeloma enzymology, Multiple Myeloma genetics, Tumor Hypoxia genetics
- Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy, which is characterized by clonal proliferation of neoplastic plasma cells in the bone marrow. This microenvironment is characterized by low oxygen levels (1-6% O
2 ), known as hypoxia. For MM cells, hypoxia is a physiologic feature that has been described to promote an aggressive phenotype and to confer drug resistance. However, studies on hypoxia are scarce and show little conformity. Here, we analyzed the mRNA expression of previously determined hypoxia markers to define the temporal adaptation of MM cells to chronic hypoxia. Subsequent analyses of the global proteome in MM cells and the stromal cell line HS-5 revealed hypoxia-dependent regulation of proteins, which directly or indirectly upregulate glycolysis. In addition, chronic hypoxia led to MM-specific regulation of nine distinct proteins. One of these proteins is the cysteine protease legumain (LGMN), the depletion of which led to a significant growth disadvantage of MM cell lines that is enhanced under hypoxia. Thus, herein, we report a methodologic strategy to examine MM cells under physiologic hypoxic conditions in vitro and to decipher and study previously masked hypoxia-specific therapeutic targets such as the cysteine protease LGMN.- Published
- 2022
- Full Text
- View/download PDF
19. High CD34 positive stem cell dosage improves thrombocyte recovery in obese myeloma patients undergoing autologous stem cell transplantation.
- Author
-
Enßle JC, Wolf S, von Metzler I, Weber S, Bialleck H, Seifried E, Serve H, Scheich S, and Steffen B
- Subjects
- Antigens, CD34, Blood Platelets, Hematopoietic Stem Cell Mobilization, Humans, Obesity complications, Obesity therapy, Transplantation, Autologous, Hematopoietic Stem Cell Transplantation, Multiple Myeloma therapy
- Published
- 2021
- Full Text
- View/download PDF
20. Clinical characteristics and outcome of multiple myeloma patients with concomitant COVID-19 at Comprehensive Cancer Centers in Germany.
- Author
-
Engelhardt M, Shoumariyeh K, Rösner A, Ihorst G, Biavasco F, Meckel K, von Metzler I, Treurich S, Hebart H, Grube M, Kull M, Bassermann F, Schäfer-Eckart K, Hoferer A, Einsele H, Rasche L, and Wäsch R
- Subjects
- Aged, Aged, 80 and over, COVID-19 transmission, COVID-19 virology, Combined Modality Therapy, Female, Follow-Up Studies, Humans, Male, Middle Aged, Multiple Myeloma therapy, Multiple Myeloma virology, Prognosis, Retrospective Studies, Survival Rate, COVID-19 complications, Multiple Myeloma mortality, Multiple Myeloma pathology, SARS-CoV-2 isolation & purification
- Published
- 2020
- Full Text
- View/download PDF
21. Enhancing the Activation and Releasing the Brakes: A Double Hit Strategy to Improve NK Cell Cytotoxicity Against Multiple Myeloma.
- Author
-
Tognarelli S, Wirsching S, von Metzler I, Rais B, Jacobs B, Serve H, Bader P, and Ullrich E
- Subjects
- Adult, Aged, Hematopoietic Stem Cell Transplantation methods, Histocompatibility Antigens Class I immunology, Humans, Interferon-gamma immunology, Interleukin-15 immunology, Interleukin-2 immunology, Middle Aged, NK Cell Lectin-Like Receptor Subfamily C immunology, Transplantation, Autologous methods, HLA-E Antigens, Cytotoxicity, Immunologic immunology, Killer Cells, Natural immunology, Lymphocyte Activation immunology, Multiple Myeloma immunology, Multiple Myeloma therapy
- Abstract
Natural killer (NK) cells are innate lymphocytes with a strong antitumor ability. In tumor patients, such as multiple myeloma (MM) patients, an elevated number of NK cells after stem cell transplantation (SCT) has been reported to be correlated with a higher overall survival rate. With the aim of improving NK cell use for adoptive cell therapy, we also addressed the cytotoxicity of patient-derived, cytokine-stimulated NK cells against MM cells at specific time points: at diagnosis and before and after autologous stem cell transplantation. Remarkably, after cytokine stimulation, the patients' NK cells did not significantly differ from those of healthy donors. In a small cohort of MM patients, we were able to isolate autologous tumor cells, and we could demonstrate that IL-2/15 stimulated autologous NK cells were able to significantly improve their killing capacity of autologous tumor cells. With the aim to further improve the NK cell killing capacity against MM cells, we investigated the potential use of NK specific check point inhibitors with focus on NKG2A because this inhibitory NK cell receptor was upregulated following ex vivo cytokine stimulation and MM cells showed HLA-E expression that could even be increased by exposure to IFN-γ. Importantly, blocking of NKG2A resulted in a significant increase in the NK cell-mediated lysis of different MM target cells. Finally, these results let suggest that combining cytokine induced NK cell activation and the specific check point inhibition of the NKG2A-mediated pathways can be an effective strategy to optimize NK cell therapeutic approaches for treatment of multiple myeloma.
- Published
- 2018
- Full Text
- View/download PDF
22. Clinical Impact of Colonization with Multidrug-Resistant Organisms on Outcome after Autologous Stem Cell Transplantation: A Retrospective Single-Center Study.
- Author
-
Scheich S, Reinheimer C, Brandt C, Wichelhaus TA, Hogardt M, Kempf VAJ, Brunnberg U, Brandts C, Ballo O, von Metzler I, Kessel J, Serve H, and Steffen B
- Subjects
- Adult, Aged, Analysis of Variance, Anti-Bacterial Agents therapeutic use, Drug Resistance, Multiple, Bacterial, Female, Gram-Negative Bacterial Infections immunology, Gram-Negative Bacterial Infections microbiology, Gram-Negative Bacterial Infections mortality, Gram-Positive Bacterial Infections immunology, Gram-Positive Bacterial Infections microbiology, Gram-Positive Bacterial Infections mortality, Hematologic Neoplasms immunology, Hematologic Neoplasms microbiology, Hematologic Neoplasms mortality, Humans, Male, Middle Aged, Myeloablative Agonists therapeutic use, Neutropenia immunology, Neutropenia microbiology, Neutropenia mortality, Retrospective Studies, Survival Analysis, Transplantation Conditioning methods, Transplantation, Autologous, Transplantation, Homologous, Treatment Outcome, Gram-Negative Bacterial Infections therapy, Gram-Positive Bacterial Infections therapy, Hematologic Neoplasms therapy, Hematopoietic Stem Cell Transplantation, Neutropenia therapy
- Abstract
A significant increase in infections caused by multidrug-resistant organisms (MDRO) has been observed in recent years, resulting in an increase of mortality in all fields of health care. Hematological patients are particularly affected by MDRO infections because of disease- and therapy-related immunosuppression. To determine the impact of colonization with MDRO on overall survival, we retrospectively analyzed data from patients undergoing autologous hematopoietic stem cell transplantation at our institution. In total, 184 patients were identified, mainly patients with lymphomas (n = 98, 53.3%), multiple myelomas (n = 80, 43.5%), germ cell cancers (n = 5, 2.7%), or acute myeloid leukemia (n = 1, .5%). Forty patients (21.7%) tested positive for MDRO colonization. At a median follow-up time of 21.5 months, the main causes of death were infection in colonized and disease progression in noncolonized patients. Nonrelapse mortality (NRM) was higher in patients who tested positive for MDRO than in the noncolonized group (25.4% versus 3%, P < .001). Interestingly, NRM in neutropenia after autologous transplantation did not differ between colonized and noncolonized patients. Colonized patients, however, had inferior overall survival after autologous transplantation in univariate (61.7% versus 73.3%, P = .005) as well as in multivariate analysis (hazard ratio, 2.463; 95% confidence interval, 1.311 to 4.626; P = .005). We conclude that the period after discharge from hospital after autologous transplantation seems critical and patients with MDRO colonization should be observed closely for infections in the post-transplantation period in outpatient care., (Copyright © 2017 The American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
23. The proteasome inhibitor bortezomib stimulates osteoblastic differentiation of human osteoblast precursors via upregulation of vitamin D receptor signalling.
- Author
-
Kaiser MF, Heider U, Mieth M, Zang C, von Metzler I, and Sezer O
- Subjects
- Base Sequence, Bortezomib, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Line, Tumor, Cells, Cultured, Coculture Techniques, Humans, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells drug effects, Mesenchymal Stem Cells metabolism, Multiple Myeloma complications, Multiple Myeloma drug therapy, Multiple Myeloma metabolism, Osteoblasts metabolism, Osteocalcin genetics, Osteoclasts cytology, Osteoclasts drug effects, Osteoclasts metabolism, Osteopontin genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Calcitriol genetics, Recombinant Proteins genetics, Recombinant Proteins metabolism, Signal Transduction drug effects, Transfection, Up-Regulation drug effects, Vitamin D Deficiency drug therapy, Vitamin D Deficiency etiology, Vitamin D Deficiency metabolism, Boronic Acids pharmacology, Osteoblasts cytology, Osteoblasts drug effects, Proteasome Inhibitors pharmacology, Pyrazines pharmacology, Receptors, Calcitriol metabolism
- Abstract
Interactions of myeloma cells with the bone marrow microenvironment lead to enhanced osteoclast recruitment and impaired osteoblast activity. Recent evidence revealed that the proteasome inhibitor bortezomib stimulates osteoblast differentiation, but the mechanisms are not fully elucidated. We hypothesised that bortezomib could influence osteoblastic differentiation via alteration of vitamin D signalling by blocking the proteasomal degradation of the vitamin D receptor (VDR). This is of clinical importance, as a high rate of vitamin D deficiency was reported in patients with myeloma. We performed cocultures of primary human mesenchymal stem cells (hMSCs) and human osteoblasts (hOBs) with myeloma cells, which resulted in an inhibition of the vitamin D-dependent differentiation of osteoblast precursors. Treatment with bortezomib led to a moderate increase in osteoblastic differentiation markers in hMSCs and hOBs. Importantly, this effect could be strikingly increased when vitamin D was added. Bortezomib led to enhanced nuclear VDR protein levels in hMSCs. Primary hMSCs transfected with a VDR luciferase reporter construct showed a strong increase in VDR signalling with bortezomib. In summary, stimulation of VDR signalling is a mechanism for the bortezomib-induced stimulation of osteoblastic differentiation. The data suggest that supplementation of vitamin D in patients with myeloma treated with bortezomib is crucial for optimal bone formation., (© 2013 John Wiley & Sons A/S.)
- Published
- 2013
- Full Text
- View/download PDF
24. The novel, orally bioavailable HSP90 inhibitor NVP-HSP990 induces cell cycle arrest and apoptosis in multiple myeloma cells and acts synergistically with melphalan by increased cleavage of caspases.
- Author
-
Lamottke B, Kaiser M, Mieth M, Heider U, Gao Z, Nikolova Z, Jensen MR, Sterz J, von Metzler I, and Sezer O
- Subjects
- Administration, Oral, Biological Availability, Enzyme Activation, Humans, Proteolysis, Pyridones administration & dosage, Pyridones pharmacokinetics, Pyrimidines administration & dosage, Pyrimidines pharmacokinetics, Apoptosis drug effects, Caspases metabolism, Cell Cycle drug effects, Melphalan pharmacology, Multiple Myeloma pathology, Pyridones pharmacology, Pyrimidines pharmacology
- Abstract
Heat shock protein 90 (HSP90) binds and stabilizes numerous proteins and kinases essential for myeloma cell survival and proliferation. We and others have recently demonstrated that inhibition of HSP90 by small molecular mass inhibitors induces cell death in multiple myeloma (MM). However, some of the HSP90 inhibitors involved in early clinical trials have shown limited antitumor activity and unfavorable toxicity profiles. Here, we analyzed the effects of the novel, orally bioavailable HSP90 inhibitor NVP-HSP990 on MM cell proliferation and survival. The inhibitor led to a significant reduction in myeloma cell viability and induced G2 cell cycle arrest, degradation of caspase-8 and caspase-3, and induction of apoptosis. Inhibition of the HSP90 ATPase activity was accompanied by the degradation of MM phospho-Akt and phospho-ERK1/2 and upregulation of Hsp70. Exposure of MM cells to a combination of NVP-HSP990 and either melphalan or histone deacetylase (HDAC) inhibitors caused synergistic inhibition of viability, increased induction of apoptosis, and was able to overcome the primary resistance of the cell line RPMI-8226 to HSP90 inhibition. Combined incubation with melphalan and NVP-HSP990 led to synergistically increased cleavage of caspase-2, caspase-9, and caspase-3. These data demonstrate promising activity for NVP-HSP990 as single agent or combination treatment in MM and provide a rationale for clinical trials., (© 2012 John Wiley & Sons A/S.)
- Published
- 2012
- Full Text
- View/download PDF
25. BSc2118 is a novel proteasome inhibitor with activity against multiple myeloma.
- Author
-
Sterz J, Jakob C, Kuckelkorn U, Heider U, Mieth M, Kleeberg L, Kaiser M, Kloetzel PM, Sezer O, and von Metzler I
- Subjects
- Antineoplastic Agents, Apoptosis drug effects, Bone Marrow Examination, Cell Cycle drug effects, Cell Line, Tumor, Cells, Cultured, Dose-Response Relationship, Drug, Humans, Leukocytes, Mononuclear drug effects, Multiple Myeloma pathology, Translocation, Genetic, Butanes pharmacology, Multiple Myeloma drug therapy, Oligopeptides pharmacology, Proteasome Inhibitors
- Abstract
Objectives: The ubiquitin-proteasome system emerged as a new therapeutic target in cancer treatment. The purpose of this study was to elucidate the effects of the novel proteasome inhibitor BSc2118 on t(4;14) positive and negative multiple myeloma (MM) cells and normal peripheral blood mononuclear cells (PBMNC)., Methods: Human MM cell lines OPM-2, RPMI-8226, and U266 and primary MM cells from bone marrow aspirates were exposed to BSc2118. Cytotoxicity levels were evaluated using the MTT-test. BSc2118-induced apoptosis was analyzed by annexin-V assay. Further methods used included proteasomal activity determination, cell cycle analysis, western blot, and transcription factor assays., Results: In OPM-2, RPMI-8226, U266 cell lines and primary MM cells, BSc2118 caused dose-dependent growth inhibitory effects. After 48 h, dose-dependent apoptosis occurred both in cell lines and primary myeloma cells irrespective of t(4;14). A significant G2-M cell cycle arrest occurred after 24 h. Furthermore, we observed a marked inhibition of intracellular proteasome activity, an increase in intracellular p21 levels, and an inhibition of NF-kappaB activation. The toxicity against PBMNC remained low, suggesting a broad therapeutic range of this agent., Conclusion: Taken together, BSc2118 shows significant antimyeloma activity and may be considered as a promising agent in cancer drug development.
- Published
- 2010
- Full Text
- View/download PDF
26. Decrease in CD4+ T-cell counts in patients with multiple myeloma treated with bortezomib.
- Author
-
Heider U, Rademacher J, Kaiser M, Kleeberg L, von Metzler I, and Sezer O
- Subjects
- Acyclovir immunology, Acyclovir pharmacology, Acyclovir therapeutic use, Boronic Acids, Bortezomib, CD4 Lymphocyte Count, CD4-Positive T-Lymphocytes virology, Herpes Zoster drug therapy, Herpes Zoster virology, Humans, Male, Multiple Myeloma immunology, Pyrazines, T-Lymphocytes immunology, T-Lymphocytes virology, CD4-Positive T-Lymphocytes immunology, Herpes Zoster immunology, Herpesvirus 3, Human drug effects, Herpesvirus 3, Human immunology, Multiple Myeloma drug therapy, Multiple Myeloma virology
- Abstract
Background: Bortezomib is highly effective in multiple myeloma and is widely used in this disease. Recently, an increased incidence of varicella zoster virus (VZV) reactivation was reported in patients with myeloma undergoing bortezomib treatment., Patients and Methods: We investigated the influence of bortezomib on T-cell subpopulations in 53 patients with myeloma before initiation of bortezomib and during therapy., Results: A decrease of CD4+ T cells was seen in 41 of 53 patients (77%). The median CD3+/CD4+ lymphocyte counts declined from 494/microL (range, 130-2187/microL) to 274/microL (range, 41-1404/microL) during bortezomib treatment (P < .001). In the majority of patients (40 of 53 patients, 75%), CD4+ lymphocytes dropped to < 400/microL during bortezomib treatment, and in 18 of 53 patients (33.9%) the CD4+ T cells fell below 200/microL. The minimum CD4S+ cell count was observed at a median of 6 weeks (range, 2-22 weeks) after initiation of treatment. The incidence of herpes zoster reactivation was 5.7% in the whole population of patients with myeloma receiving bortezomib. Nineteen of 53 patients received acyclovir at a dose of 400 mg daily as prophylaxis against VZV reactivation. In this group, none of the patients developed herpes zoster. The incidence of VZV reactivation in patients not receiving acyclovir was 3 of 34 (8.8%). Importantly, occurrence of herpes zoster was associated with reduced CD4+ T-cell subpopulation: all patients who developed herpes zoster had CD4+ lymphocytes < 400/microL., Conclusion: Our results show that bortezomib leads to a transient decrease in CD4+ lymphocytes, accompanied by an increased incidence of VZV infections. The antiviral prophylaxis with acyclovir is effective in patients with myeloma treated with bortezomib.
- Published
- 2010
- Full Text
- View/download PDF
27. Synergistic action of the novel HSP90 inhibitor NVP-AUY922 with histone deacetylase inhibitors, melphalan, or doxorubicin in multiple myeloma.
- Author
-
Kaiser M, Lamottke B, Mieth M, Jensen MR, Quadt C, Garcia-Echeverria C, Atadja P, Heider U, von Metzler I, Türkmen S, and Sezer O
- Subjects
- Antibiotics, Antineoplastic, Antineoplastic Agents, Alkylating, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Doxorubicin agonists, Drug Evaluation, Preclinical, Drug Synergism, HSP90 Heat-Shock Proteins metabolism, Histone Deacetylase Inhibitors agonists, Histone Deacetylases metabolism, Humans, Isoxazoles agonists, Isoxazoles therapeutic use, Melphalan agonists, Multiple Myeloma metabolism, Resorcinols agonists, Resorcinols therapeutic use, Doxorubicin pharmacology, HSP90 Heat-Shock Proteins antagonists & inhibitors, Histone Deacetylase Inhibitors pharmacology, Isoxazoles pharmacology, Melphalan pharmacology, Multiple Myeloma drug therapy, Resorcinols pharmacology
- Abstract
Heat shock protein 90 (HSP90) is a promising target for tumor therapy. The novel HSP90 inhibitor NVP-AUY922 has preclinical activity in multiple myeloma, however, little is known about effective combination partners to design clinical studies. Multiple myeloma cell lines, OPM-2, RPMI-8226, U-266, LP-1, MM1.S, and primary myeloma cells were exposed to NVP-AUY922 and one of the combination partners histone deacetylase inhibitor NVP-LBH589, suberoylanilide hydroxamic acid (SAHA), melphalan, or doxorubicin, either simultaneously or in sequential patterns. Effects on cell proliferation and apoptosis were determined. Synergistic effects were evaluated using the method of Chou and Talalay. Combined sequential incubation with NVP-AUY922 and SAHA showed that best synergistic effects were achieved with 24 h preincubation with SAHA followed by another 48 h of combination treatment. Combination of NVP-AUY922 with SAHA, NVP-LBH589, melphalan, or doxorubicin resulted in synergistic inhibition of viability, with strong synergy (combination index < 0.3) in the case of melphalan. Importantly, resistance of the RPMI-8226 cell line and relative resistance of some primary myeloma cells against NVP-AUY922 could be overcome by combination treatment. These data show impressive synergistic action of the novel HSP90 inhibitor NVP-AUY922 with melphalan, doxorubicin, NVP-LBH589, and SAHA in multiple myeloma and build the frame work for clinical trials.
- Published
- 2010
- Full Text
- View/download PDF
28. Synergistic interaction of proteasome and topoisomerase II inhibition in multiple myeloma.
- Author
-
von Metzler I, Heider U, Mieth M, Lamottke B, Kaiser M, Jakob C, and Sezer O
- Subjects
- Antineoplastic Agents pharmacology, Boronic Acids pharmacology, Bortezomib, Cell Cycle drug effects, Cell Cycle physiology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Cell Survival physiology, DNA Topoisomerases, Type II metabolism, Drug Synergism, Etoposide pharmacology, Humans, Multiple Myeloma enzymology, NF-kappa B drug effects, NF-kappa B metabolism, Protease Inhibitors pharmacology, Proteasome Endopeptidase Complex metabolism, Proto-Oncogene Proteins c-bcl-2 drug effects, Proto-Oncogene Proteins c-bcl-2 metabolism, Pyrazines pharmacology, Antineoplastic Agents therapeutic use, Boronic Acids therapeutic use, Etoposide therapeutic use, Multiple Myeloma drug therapy, Protease Inhibitors therapeutic use, Proteasome Inhibitors, Pyrazines therapeutic use, Topoisomerase II Inhibitors
- Abstract
Multiple myeloma is a malignancy of terminally differentiated plasma cells and is incurable in the majority of the patients. Thus, novel effective treatment regimens are urgently needed. In this study, we examined the effects of co-treatment with proteasome-inhibitor bortezomib and topoisomerase II inhibitor etoposide in multiple myeloma cells lines OPM-2, RPMI-S and NCI-H929. Using the median effect method of Chou and Talalay, we evaluated the combination indices (CI) for simultaneous and sequential treatment schedules. In the sequential treatment schedule, we found strong synergistic effects in all three cell lines, even at low single-agent cytotoxicity levels. When cells were treated simultaneously with both drugs, the synergy was present but less pronounced than in the sequential treatment schedule. The synergistic effects observed in the co-treatment schedules were accompanied by an inhibition of anti-apoptotic effects that were induced by etoposide alone. Namely, bortezomib abrogated both etoposide-induced NF-kappaB activation and etoposide-induced bcl-2 up-regulation. Our data suggest that combining etoposide with bortezomib might be useful for cancer treatment, as bortezomib potentially inhibits counter-regulatory mechanisms of tumor cells, which are induced by topoisomerase II inhibition and which may contribute to acquired chemoresistance.
- Published
- 2009
- Full Text
- View/download PDF
29. Synergistic interaction of the histone deacetylase inhibitor SAHA with the proteasome inhibitor bortezomib in cutaneous T cell lymphoma.
- Author
-
Heider U, Rademacher J, Lamottke B, Mieth M, Moebs M, von Metzler I, Assaf C, and Sezer O
- Subjects
- Apoptosis, Boronic Acids pharmacokinetics, Bortezomib, Cell Line, Tumor, Cell Survival, Dose-Response Relationship, Drug, Drug Synergism, Histone Deacetylase Inhibitors, Humans, Hydroxamic Acids pharmacokinetics, Pharmacokinetics, Pyrazines pharmacokinetics, Reactive Oxygen Species analysis, Vorinostat, Boronic Acids pharmacology, Hydroxamic Acids pharmacology, Lymphoma, T-Cell, Cutaneous drug therapy, Pyrazines pharmacology
- Abstract
Proteasome inhibitors and histone deacetylase (HDAC) inhibitors are novel targeted therapies being evaluated in clinical trials for cutaneous T-cell lymphoma (CTCL). However, data in regard to tumor biology are limited with these agents. In the present study we analyzed the effects of the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) and the proteasome inhibitor bortezomib on human CTCL cells. Four CTCL cell lines (SeAx, Hut-78, MyLa, and HH) were exposed to bortezomib and/ or SAHA at different concentrations. Cell viability was quantified using the MTT assay. In addition, apoptosis and generation of reactive oxygen species were analyzed. Both agents potently inhibited cell viability and induced apoptosis. After 48 h of incubation, IC50 of bortezomib was noted at 8.3 nm, 7.9 nm, 6.3 nm, and 22.5 nm in SeAx, Hut-78, HH, and MyLa cells, respectively. For SAHA, the IC50 values were at 0.6 microm in SeAx cells, 0.75 microm in Hut-78 cells, 0.9 microm in HH cells, and 4.4 microm in MyLa cells. Importantly, combined treatment resulted in synergistic cytotoxic effects, as indicated by Combination indices values <1 using the median effect method of Chou and Talalay. We furthermore found that combined treatment with both agents lead to a decreased proteasome activity, an upregulation of the cell regulators p21 and p27 and increased expression of phosphorylated p38. In addition, we showed that SAHA reduced the vascular endothelial growth factor production of CTCL cells. Our results demonstrate that bortezomib and SAHA synergistically induce apoptosis in CTCL cells and thus provide a rationale for clinical trials of combined proteasome and histone deacetylase inhibition in the treatment of CTCL.
- Published
- 2009
- Full Text
- View/download PDF
30. Curcumin diminishes human osteoclastogenesis by inhibition of the signalosome-associated I kappaB kinase.
- Author
-
von Metzler I, Krebbel H, Kuckelkorn U, Heider U, Jakob C, Kaiser M, Fleissner C, Terpos E, and Sezer O
- Subjects
- Bone Diseases etiology, Bone Diseases prevention & control, Bone Resorption prevention & control, Cell Differentiation drug effects, Cell Survival drug effects, Enzyme-Linked Immunosorbent Assay, Humans, Neoplasms complications, Osteoclasts drug effects, Osteoclasts pathology, Reference Values, Transcription Factors analysis, Antineoplastic Agents therapeutic use, Curcumin therapeutic use, I-kappa B Kinase antagonists & inhibitors, Osteoclasts cytology
- Abstract
Purpose: Curcumin is a natural polyphenolic derogate extracted from spice turmeric, exhibiting anti-inflammatory and chemopreventive activities. It was described to interact with the signalosome-associated kinases and the proteasome-ubiquitin system, which both are involved in the osteoclastogenesis. Thus, we hypothesized that curcumin could diminish osteoclast differentiation and function., Methods: For the experiments considering osteoclast differentiation and resorptional activities, preosteoclasts were cultured for 4 weeks and treated with curcumin at subapoptotic dosages. Derived mature osteoclasts were identified as large, multinucleated cells with expression of tartrate-resistant acid phosphatase activity. Formation of resorption lacunae, a hallmark of osteoclast activity, was quantified using dentine pits and light microscopy. The signaling pathways were examined by ELISA-based methods and by immunoblotting., Results: Both 1 and 10 microM curcumin abrogated osteoclast differentiation (by 56 and 81%) and function (by 56 and 99%) (P < 0.05) dose-dependently. The effects were accompanied by the inhibition of I kappaB phosphorylation and NF-kappaB activation. In contrast, subtoxic doses did not have any significant effects on proteasome inhibition., Conclusion: This manuscript is the first report that describes the effects of curcumin toward human osteoclastogenesis, and builds the framework for clinical trials of curcumin in the treatment of cancer-induced lytic bone disease.
- Published
- 2009
- Full Text
- View/download PDF
31. The potential of proteasome inhibitors in cancer therapy.
- Author
-
Sterz J, von Metzler I, Hahne JC, Lamottke B, Rademacher J, Heider U, Terpos E, and Sezer O
- Subjects
- Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Boronic Acids administration & dosage, Boronic Acids adverse effects, Boronic Acids pharmacology, Boronic Acids therapeutic use, Bortezomib, Cell Line, Tumor drug effects, Clinical Trials as Topic statistics & numerical data, Combined Modality Therapy, Drug Design, Drug Screening Assays, Antitumor, Hematologic Neoplasms drug therapy, Hematologic Neoplasms metabolism, Hematologic Neoplasms surgery, Hematopoietic Stem Cell Transplantation, Humans, Neoplasm Proteins metabolism, Neoplasms enzymology, Protease Inhibitors administration & dosage, Protease Inhibitors adverse effects, Protease Inhibitors pharmacology, Protein Processing, Post-Translational drug effects, Protein Processing, Post-Translational physiology, Pyrazines administration & dosage, Pyrazines adverse effects, Pyrazines pharmacology, Pyrazines therapeutic use, Salvage Therapy, Ubiquitin physiology, Ubiquitination, Antineoplastic Agents therapeutic use, Neoplasm Proteins antagonists & inhibitors, Neoplasms drug therapy, Protease Inhibitors therapeutic use, Proteasome Inhibitors
- Abstract
Background: The ubiquitin-proteasome system has become a promising novel molecular target in cancer due to its critical role in cellular protein degradation, its interaction with cell cycle and apoptosis regulation and its unique mechanism of action., Objective: This review focuses both on preclinical results and on data from clinical trials with proteasome inhibitors in cancer., Methods: Results in hematological malignancies and solid tumors were included, and important data presented in abstract form were considered in this review., Results/conclusion: Bortezomib as first-in-class proteasome inhibitor has proven to be highly effective in some hematological malignancies, overcomes conventional chemoresistance, directly induces cell cycle arrest and apoptosis, and also targets the tumor microenvironment. It has been granted approval by the FDA for relapsed multiple myeloma, and recently for relapsed mantle cell lymphoma. Combination chemotherapy regimens have been developed providing high remission rates and remission quality in frontline treatment or in the relapsed setting in multiple myeloma. The combination of proteasome inhibition with novel targeted therapies is an emerging field in oncology. Moreover, novel proteasome inhibitors, such as NPI-0052 and carfilzomib, have been developed. This review summarizes our knowledge of the ubiquitin-proteasome system and recent data from cancer clinical trials.
- Published
- 2008
- Full Text
- View/download PDF
32. Interactions of myeloma cells with osteoclasts promote tumour expansion and bone degradation through activation of a complex signalling network and upregulation of cathepsin K, matrix metalloproteinases (MMPs) and urokinase plasminogen activator (uPA).
- Author
-
Hecht M, von Metzler I, Sack K, Kaiser M, and Sezer O
- Subjects
- Cathepsin K, Cell Communication, Cell Line, Tumor, Coculture Techniques, Gene Expression Regulation, Neoplastic, Humans, Multiple Myeloma complications, Osteoclasts pathology, Tartrate-Resistant Acid Phosphatase, Tumor Cells, Cultured, Up-Regulation genetics, Acid Phosphatase genetics, Bone Resorption etiology, Cathepsins genetics, Cell Proliferation, Isoenzymes genetics, Matrix Metalloproteinases genetics, Multiple Myeloma pathology, Signal Transduction, Urokinase-Type Plasminogen Activator genetics
- Abstract
Bone destruction is one of the most debilitating manifestations of multiple myeloma (MM) and results from the interaction of myeloma cells with the bone marrow microenvironment. Within the bone marrow, the disturbed balance between osteoclasts and osteoblasts is important for the development of lytic lesions. However, the mechanisms behind myeloma-mediated bone destruction are not completely understood. In order to address the importance of myeloma cell-osteoclast interactions in MM pathogenesis, we have developed a functional coculture system. We found that myeloma-osteoclast interactions resulted in stimulation of myeloma cell growth and osteoclastic activity through activation of major signalling pathways and upregulation of proteases. Signals from osteoclasts activated the p44/p42 MAPK, STAT3 and PI3K/Akt pathways in myeloma cells. In turn, myeloma cells triggered p38 MAPK and NF-kappaB signalling in osteoclasts. Myeloma-osteoclast interactions stimulated the production of TRAP, cathepsin K, matrix metalloproteinase (MMP)-1, -9, and urokinase plasminogen activator (uPA). Consistent data with myeloma cell lines and primary myeloma cells underlined the biological relevance of these findings. In conclusion, we demonstrated the critical role of myeloma cell-osteoclast interactions in the existing interdependence between tumour expansion and bone disease. The identified molecular events might provide the rationale for novel treatment strategies.
- Published
- 2008
- Full Text
- View/download PDF
33. Synergistic interaction of the histone deacetylase inhibitor SAHA with the proteasome inhibitor bortezomib in mantle cell lymphoma.
- Author
-
Heider U, von Metzler I, Kaiser M, Rosche M, Sterz J, Rötzer S, Rademacher J, Jakob C, Fleissner C, Kuckelkorn U, Kloetzel PM, and Sezer O
- Subjects
- Apoptosis, Boronic Acids chemistry, Bortezomib, Caspases metabolism, Cell Line, Tumor, Cell Survival, Drug Synergism, Enzyme Inhibitors pharmacology, Humans, Hydroxamic Acids chemistry, NF-kappa B metabolism, Pyrazines chemistry, Reactive Oxygen Species, Treatment Outcome, Vorinostat, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Boronic Acids pharmacology, Histone Deacetylase Inhibitors, Hydroxamic Acids pharmacology, Lymphoma, Mantle-Cell drug therapy, Proteasome Inhibitors, Pyrazines pharmacology
- Abstract
Objectives: Mantle cell lymphoma (MCL) is an incurable B cell lymphoma, and novel treatment strategies are urgently needed. We evaluated the effects of combined treatment with the proteasome inhibitor bortezomib and the histone deacetylase inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA) on MCL. Bortezomib acts by targeting the proteasome, and--among other mechanisms--results in a reduced nuclear factor-kappa B (NF-kappaB) activity. HDACi promote histone acetylation, and also interfere with NF-kappaB signaling., Methods: Human MCL cell lines (JeKo-1, Granta-519 and Hbl-2) were exposed to bortezomib and/or SAHA. Cell viability and apoptosis were quantified by the MTT and annexin-V assay, respectively. Reactive oxygen species (ROS) were analyzed using the fluorophore H2DCFDA. In addition, activated caspases, proteasome- and NF-kappaB activity were quantified., Results: Combined incubation with bortezomib and SAHA resulted in synergistic cytotoxic effects, as indicated by combination index values <1 using the median effect method of Chou and Talalay. The combination of both inhibitors led to a strong increase in apoptosis as compared to single agents and was accompanied by enhanced ROS generation, while each agent alone only modestly induced ROS. The free radical scavenger N-acetyl-L-cysteine blocked the ROS generation and reduced the apoptosis significantly. In addition, coexposure of bortezomib and SAHA led to increased caspase-3, -8 and -9 activity, marked reduction of proteasome activity and decrease of NF-kappaB activity., Conclusions: This is the first report giving evidence that SAHA and bortezomib synergistically induce apoptosis in MCL cells. These data build the framework for clinical trials using combined proteasome and histone deacetylase inhibition in the treatment of MCL.
- Published
- 2008
- Full Text
- View/download PDF
34. Osteoblasts promote migration and invasion of myeloma cells through upregulation of matrix metalloproteinases, urokinase plasminogen activator, hepatocyte growth factor and activation of p38 MAPK.
- Author
-
Hecht M, Heider U, Kaiser M, von Metzler I, Sterz J, and Sezer O
- Subjects
- Cell Communication, Cell Line, Tumor, Cell Movement, Coculture Techniques, Collagen, Collagen Type I metabolism, Culture Media, Conditioned chemistry, Drug Combinations, Hepatocyte Growth Factor analysis, Humans, Laminin, Matrix Metalloproteinase 1 analysis, Matrix Metalloproteinase 1 metabolism, Matrix Metalloproteinase 2 analysis, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinases analysis, Multiple Myeloma metabolism, Neoplasm Invasiveness, Proteoglycans, Up-Regulation, Urokinase-Type Plasminogen Activator analysis, Hepatocyte Growth Factor metabolism, MAP Kinase Signaling System physiology, Matrix Metalloproteinases metabolism, Multiple Myeloma pathology, Osteoblasts physiology, Urokinase-Type Plasminogen Activator metabolism
- Abstract
Formation of osteolytic lesions is a key pathophysiological feature in multiple myeloma and results from the interaction of myeloma cells with the bone marrow microenvironment. Matrix metalloproteinases (MMPs) and plasmin may be involved in bone destruction, but their precise roles have not been clarified. Furthermore, the impact of osteoblast-related alterations on myeloma bone disease is not well understood. We addressed this complex phenomenon by applying a coculture system between myeloma cells and osteoblasts. Osteoblasts induced expression of MMP-1 and upregulated the expression of MMP-2, urokinase plasminogen activator (uPA) and hepatocyte growth factor (HGF) in myeloma cells. In turn, interaction with myeloma cells led to abundant MMP-1 expression in osteoblasts. Because MMP-1 degrades collagen, its upregulation might represent an essential mechanism contributing to bone destruction. Cocultures using primary myeloma cells confirmed the results obtained with cell lines. The mechanisms responsible for MMP-1 upregulation are mediated by both membrane-bound and soluble factors, and involve the p38 mitogen-activated protein kinase (MAPK) pathway. The interaction with osteoblasts enhances the capability of myeloma cells to transmigrate and invade through Matrigel or type I collagen. Using appropriate inhibitors, we provide evidence that these processes involve MMPs, uPA, HGF and activation of p38 MAPK.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.